1
|
Singh D, Kumar S, Mishra R, Anjali, Tripathi RK, Sachdev M. HIV1-Nef perturbs the integrity of blood testis barrier in rat model. Tissue Barriers 2025; 13:2357406. [PMID: 38778621 DOI: 10.1080/21688370.2024.2357406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The blood-testis barrier is a specialized feature within the mammalian testis, located in close proximity to the basement membrane of seminiferous tubules. This barrier serves to divide the seminiferous epithelium into distinct basal and adluminal (apical) compartments. The selectivity of the BTB to foreign particles makes it a safe haven for the virus, and the high affinity of HIV for testis might lead to the vertical transmission of the virus. In the present study, recombinant HIV1-Nef (rNef) protein was injected intravenously to examine the effect of rNef on BTB. SD male rats received 250 µg and 500 µg of rNef along with 2% Evans blue dye within 1 ml through the tail vein. After 1 hour of perfusion, the animals were sacrificed for analysis. The dye migration assay and ELISA confirmed a significant impairment in the blood-testis barrier (BTB) and the manifestation of rNef in testes tissues, respectively. Moreover, a decline in the expression of tight junction proteins, including ZO1 and Occludin, was observed during rNef-induced BTB disruption. Overall, our findings demonstrated that rNef induces BTB disruption through various signaling events. At the site of ectoplasmic specialization of the seminiferous epithelium, the localization of cadherins was found to be disrupted, making the testis a vulnerable site. In conclusion, rNef perturbs the integrity of the blood-testis barrier in rat models; hence, it can also serve as a suitable model for studying the dynamics of the blood-testis barrier.
Collapse
Affiliation(s)
- Deependra Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Saurabh Kumar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajnikant Mishra
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Anjali
- Division of Virus Research and Therapeutics, CSIR-Central Drug Research Institute, Lucknow, India
| | - R K Tripathi
- Division of Virus Research and Therapeutics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Monika Sachdev
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
2
|
Zhang Q, Ruan H, Wang X, Luo A, Ran X. Ulinastatin attenuated cardiac ischaemia/reperfusion injury by suppressing activation of the tissue kallikrein-kinin system. Br J Pharmacol 2024; 181:4988-5008. [PMID: 39294926 DOI: 10.1111/bph.16477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/29/2024] [Accepted: 05/13/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Ulinastatin has beneficial effects in patients undergoing coronary artery bypass grafting (CABG) surgery due to its anti-inflammatory properties, but the underlying mechanism remains unclear. EXPERIMENTAL APPROACH We used samples from patients undergoing CABG, a model of cardiac ischaemia-reperfusion injury (IRI) in mice and murine cardiac endothelial cell cultures to investigate links between ulinastatin, the kallikrein-kinin system (KKS), endothelial dysfunction and cardiac inflammation in the response to ischaemia/reperfusion injury (IRI). These links were assessed using clinical investigations, in vitro and in vivo experiments and RNA sequencing analysis. KEY RESULTS Ulinastatin inhibited the activity of tissue kallikrein, a key enzyme of the KKS, at 24 h after CABG surgery, which was verified in our murine cardiac ischaemia-reperfusion model. Under normal conditions, ulinastatin only inhibited kallikrein activity but did not affect bradykinin (B1/B2) receptors. Ulinastatin protected against IRI, in vivo and in vitro, by suppressing activation of the kallikrein-kinin system and down-regulating B1/B2 receptor-related signalling pathways including ERK/ iNOS, which resulted in enhanced endothelial barrier function, mitigation of inflammation and oedema, decreased infarct size, improved cardiac function and decreased mortality. Inhibition of kallikrein and knockdown of B1, but not B2 receptors prevented ERK translocation into the nucleus, reducing reperfusion-induced injury in murine cardiac endothelial cells. CONCLUSIONS AND IMPLICATIONS Treatment with ulinastatin exerts a protective influence on cardiac reperfusion by suppressing activation of the kallikrein-kinin system. Our findings highlight the potential of targeting kallikrein /bradykinin receptors to alleviate endothelial dysfunction, thus improving cardiac IRI.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Ruan
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Ran
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Milyutina YP, Kerkeshko GO, Vasilev DS, Zalozniaia IV, Bochkovskii SK, Tumanova NL, Shcherbitskaia AD, Mikhel AV, Tolibova GH, Arutjunyan AV. Placental Transport of Amino Acids in Rats with Methionine-Induced Hyperhomocysteinemia. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1711-1726. [PMID: 39523111 DOI: 10.1134/s0006297924100055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/11/2024] [Accepted: 05/02/2024] [Indexed: 11/16/2024]
Abstract
Maternal hyperhomocysteinemia (HHcy) is a risk factor for intrauterine growth restriction presumably caused by a decrease in the placental transport of nutrients. We investigated the effect of experimental HHcy induced by daily methionine administration to pregnant rats on the free amino acid levels in the maternal and fetal blood, as well as on morphological and biochemical parameters associated with the amino acid transport through the placenta. HHcy caused an increase in the levels of most free amino acids in the maternal blood on gestational day 20, while the levels of some amino acids in the fetal blood were decreased. In rats with HHcy, the maternal sinusoids in the placental labyrinth were narrowed, which was accompanied by aggregation of red blood cells. We also observed an increase in the neutral amino acid transporters (LAT1, SNAT2) protein levels and activation of 4E-BP1, a downstream effector of mTORC1 complex, in the labyrinth zone. Maternal HHcy affected the placental barrier permeability, as evidenced by intensification of the mother-to-fetus transfer of Evans Blue dye. The imbalance in the free amino acid levels in the maternal and fetal blood in HHcy may be due to the competition of homocysteine with other amino acids for common transporters, as well as a decrease in the area of exchange zone between maternal and fetal circulations in the placental labyrinth. Upregulation of the neutral amino acid transporter expression in the labyrinth zone may be a compensatory response to an insufficient intrauterine amino acid supply and fetal growth restriction.
Collapse
Affiliation(s)
- Yulia P Milyutina
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia.
| | - Gleb O Kerkeshko
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Dmitrii S Vasilev
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Irina V Zalozniaia
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Sergey K Bochkovskii
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Natalia L Tumanova
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Anastasiia D Shcherbitskaia
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Anastasiia V Mikhel
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Gulrukhsor H Tolibova
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Alexander V Arutjunyan
- D. O. Ott Research Institute of Obstetrics, Gynecology, and Reproductive Medicine, St. Petersburg, 199034, Russia
| |
Collapse
|
4
|
Parveen S, Bhat CV, Sagilkumar AC, Aziz S, Arya J, Dutta A, Dutta S, Show S, Sharma K, Rakshit S, Johnson JB, Nongthomba U, Banerjee A, Subramanian K. Bacterial pore-forming toxin pneumolysin drives pathogenicity through host extracellular vesicles released during infection. iScience 2024; 27:110589. [PMID: 39211544 PMCID: PMC11357855 DOI: 10.1016/j.isci.2024.110589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Streptococcus pneumoniae is a global priority respiratory pathogen that kills over a million people annually. The pore-forming cytotoxin, pneumolysin (PLY) is a major virulence factor. Here, we found that recombinant PLY as well as wild-type pneumococcal strains, but not the isogenic PLY mutant, upregulated the shedding of extracellular vesicles (EVs) harboring membrane-bound toxin from human THP-1 monocytes. PLY-EVs induced cytotoxicity and hemolysis dose-dependently upon internalization by recipient monocyte-derived dendritic cells. Proteomics analysis revealed that PLY-EVs are selectively enriched in key inflammatory host proteins such as IFI16, NLRC4, PTX3, and MMP9. EVs shed from PLY-challenged or infected cells induced dendritic cell maturation and primed them to infection. In vivo, zebrafish administered with PLY-EVs showed pericardial edema and mortality. Adoptive transfer of bronchoalveolar-lavage-derived EVs from infected mice to healthy recipients induced lung damage and inflammation in a PLY-dependent manner. Our findings identify that host EVs released during infection mediate pneumococcal pathogenesis.
Collapse
Affiliation(s)
- Saba Parveen
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Chinmayi V Bhat
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Aswathy C Sagilkumar
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
- Regional Centre for Biotechnology, Faridabad 121001, India
| | - Shaheena Aziz
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - J Arya
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Asmita Dutta
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Somit Dutta
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Sautan Show
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Kuldeep Sharma
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sumit Rakshit
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - John Bernet Johnson
- Virology Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Upendra Nongthomba
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Anirban Banerjee
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Karthik Subramanian
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
- Regional Centre for Biotechnology, Faridabad 121001, India
| |
Collapse
|
5
|
Mierzejewski B, Różycka J, Stremińska W, Brągiel-Pieczonka A, Sidor K, Hoser G, Bartoszewicz Z, Gewartowska M, Frontczak-Baniewicz M, Ciemerych MA, Brzóska E, Skirecki T. The Role of Pericytes in Lipopolysaccharide-Induced Murine Acute Respiratory Distress Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1443-1457. [PMID: 38705380 DOI: 10.1016/j.ajpath.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 05/07/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a heterogeneous clinical syndrome that is most commonly triggered by infection-related inflammation. Lung pericytes can respond to infection and act as immune and proangiogenic cells; moreover, these cells can differentiate into myofibroblasts in nonresolving ARDS and contribute to the development of pulmonary fibrosis. Here, we aimed to characterize the role of lung cells, which present characteristics of pericytes, such as peri-endothelial location and expression of a panel of specific markers. A murine model of lipopolysaccharide (LPS)-induced resolving ARDS was used to study their role in ARDS. The development of ARDS was confirmed after LPS instillation, which was resolved 14 days after onset. Immunofluorescence and flow cytometry showed early expansion of neural-glial antigen 2+ β-type platelet-derived growth factor receptor+ pericytes in murine lungs with loss of CD31+ β-type platelet-derived growth factor receptor+ endothelial cells. These changes were accompanied by specific changes in lung structure and loss of vascular integrity. On day 14 after ARDS onset, the composition of pericytes and endothelial cells returned to baseline values. LPS-induced ARDS activated NOTCH signaling in lung pericytes, the inhibition of which during LPS stimulation reduced the expression of its downstream target genes, pericyte markers, and angiogenic factors. Together, these data indicate that lung pericytes in response to inflammatory injury activate NOTCH signaling that supports their maintenance and in turn can contribute to recovery of the microvascular endothelium.
Collapse
Affiliation(s)
- Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Justyna Różycka
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Aneta Brągiel-Pieczonka
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Karolina Sidor
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Grażyna Hoser
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Zbigniew Bartoszewicz
- Department of Internal Diseases and Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Gewartowska
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | | | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Edyta Brzóska
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
6
|
Yamaki K, Matsuda A, Koyama Y. Importance of IgE-Induced Unique Plasma Leakage in the Skin for Urticaria-Like Symptoms in an Anaphylaxis-Dependent Spotted Distribution of Immune Complex in Skin (ASDIS) Mouse Model. Int Arch Allergy Immunol 2024; 185:1139-1153. [PMID: 38934152 DOI: 10.1159/000539215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/02/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Allergic diseases, such as anaphylaxis and urticaria, pose significant health concerns. The quest for improved prognostic outcomes in these diseases necessitates the exploration of novel therapeutic avenues. To address this need, we have developed a novel mouse model of anaphylaxis, denoted as anaphylaxis-dependent spotted distribution of immune complex in skin (ASDIS). ASDIS manifests as distinct dotted symptoms in the skin, detectable through in vivo imaging, resembling urticarial symptoms. In this study, we investigated the potential underlying mechanisms giving rise to these dotted symptoms, exploring the role of vascular permeability and characterizing the ASDIS model as a new urticaria model. METHODS We employed haired and hairless HR mice (BALB/c background) and hairless HR-1 mice (a commercially available hairless strain with an unidentified genetic background). ASDIS was induced by the simultaneous intravenous injection of anti-ovalbumin IgE and fluorescein isothiocyanate (FITC)-ovalbumin, along with Evans blue - a recognized vascular permeability indicator. Anaphylaxis and scratching behavior were monitored through rectal temperature decrease and optical observation, respectively. Histamine, platelet-activating factor, and compound 48/80 were injected with or without FITC-ovalbumin for comparative analysis. The effects of an α1 adrenergic receptor agonist applied to the skin were also examined. RESULTS In hairless mice, the simultaneous injection of histamine, compound 48/80, or IgE with FITC-ovalbumin induced comparable rectal temperature decreases and vascular permeability. However, only the combination of FITC-ovalbumin and IgE triggered ASDIS, specifically the dotted urticaria-like symptom. Evans blue visualization and optical observation of dotted swelling confirmed that the vascular permeability mediated the phenomenon. Hairless mice exhibited a more pronounced temperature decrease than their haired counterparts when exposed to histamine, platelet-activating factor, compound 48/80, and IgE with FITC-ovalbumin. The application of an α1 adrenergic receptor agonist to the skin attenuated the topical urticaria-like symptom. CONCLUSION Our experiments revealed four findings. The first is that ASDIS mirrors urticaria-like symptoms resulting from increased vascular permeability, akin to human urticaria. The second finding is that the development of dotted symptoms involves an IgE-induced, yet unidentified, mechanism not triggered by histamine or compound 48/80 alone. The third finding highlights the heightened susceptibility of hairless mice to ASDIS induction. The fourth finding demonstrates that the inhibition of ASDIS by the topical application of an α1 adrenergic receptor agonist hints at a potential anti-urticarial application for this vasoconstrictor. Further elucidation of these unidentified IgE-dependent mechanisms and the specific generation of dotted symptoms by IgE-immune complexes could provide novel insights into allergic response processes and therapeutic interventions for these conditions.
Collapse
Affiliation(s)
- Kouya Yamaki
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Akari Matsuda
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
7
|
Kamiya K, Hatayama N, Tawada M, Asai A, Yamauchi M, Kinashi H, Kunoki S, Yamaguchi M, Mizuno M, Suzuki Y, Banshodani M, Ishimoto T, Naito M, Kawanishi H, Ito Y. Role of endothelial hyaluronan in peritoneal membrane transport and disease conditions during peritoneal dialysis. Sci Rep 2024; 14:7412. [PMID: 38548914 PMCID: PMC10978880 DOI: 10.1038/s41598-024-58148-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 03/26/2024] [Indexed: 04/01/2024] Open
Abstract
Peritoneal membrane dysfunction in peritoneal dialysis (PD) is primarily attributed to angiogenesis; however, the integrity of vascular endothelial cells can affect peritoneal permeability. Hyaluronan, a component of the endothelial glycocalyx, is reportedly involved in preventing proteinuria in the normal glomerulus. One hypothesis suggests that development of encapsulating peritoneal sclerosis (EPS) is triggered by protein leakage due to vascular endothelial injury. We therefore investigated the effect of hyaluronan in the glycocalyx on peritoneal permeability and disease conditions. After hyaluronidase-mediated degradation of hyaluronan on the endothelial cells of mice, macromolecules, including albumin and β2 microglobulin, leaked into the dialysate. However, peritoneal transport of small solute molecules was not affected. Pathologically, hyaluronan expression was diminished; however, expression of vascular endothelial cadherin and heparan sulfate, a core protein of the glycocalyx, was preserved. Hyaluronan expression on endothelial cells was studied using 254 human peritoneal membrane samples. Hyaluronan expression decreased in patients undergoing long-term PD treatment and EPS patients treated with conventional solutions. Furthermore, the extent of hyaluronan loss correlated with the severity of vasculopathy. Hyaluronan on endothelial cells is involved in the peritoneal transport of macromolecules. Treatment strategies that preserve hyaluronan in the glycocalyx could prevent the leakage of macromolecules and subsequent related complications.
Collapse
Affiliation(s)
- Keisuke Kamiya
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
| | - Naoyuki Hatayama
- Department of Anatomy, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Mitsuhiro Tawada
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akimasa Asai
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
| | - Mai Yamauchi
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
| | - Hiroshi Kinashi
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
| | - Shunnosuke Kunoki
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
- Department of Nephrology, Nippon Medical School, Tokyo, Japan
| | - Makoto Yamaguchi
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
| | - Masashi Mizuno
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Yasuhiro Suzuki
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masataka Banshodani
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Takuji Ishimoto
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan
| | - Munekazu Naito
- Department of Anatomy, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Hideki Kawanishi
- Department of Surgery and Artificial Organs, Akane-Foundation, Tsuchiya General Hospital, Hiroshima, Japan
| | - Yasuhiko Ito
- Department of Nephrology and Rheumatology, Aichi Medical University, 1-1 Karimata, Yazako, Nagakute City, Aichi, 480-1195, Japan.
| |
Collapse
|
8
|
Chaube B, Citrin KM, Sahraei M, Singh AK, de Urturi DS, Ding W, Pierce RW, Raaisa R, Cardone R, Kibbey R, Fernández-Hernando C, Suárez Y. Suppression of angiopoietin-like 4 reprograms endothelial cell metabolism and inhibits angiogenesis. Nat Commun 2023; 14:8251. [PMID: 38086791 PMCID: PMC10716292 DOI: 10.1038/s41467-023-43900-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Angiopoietin-like 4 (ANGPTL4) is known to regulate various cellular and systemic functions. However, its cell-specific role in endothelial cells (ECs) function and metabolic homeostasis remains to be elucidated. Here, using endothelial-specific Angptl4 knock-out mice (Angptl4iΔEC), and transcriptomics and metabolic flux analysis, we demonstrate that ANGPTL4 is required for maintaining EC metabolic function vital for vascular permeability and angiogenesis. Knockdown of ANGPTL4 in ECs promotes lipase-mediated lipoprotein lipolysis, which results in increased fatty acid (FA) uptake and oxidation. This is also paralleled by a decrease in proper glucose utilization for angiogenic activation of ECs. Mice with endothelial-specific deletion of Angptl4 showed decreased pathological neovascularization with stable vessel structures characterized by increased pericyte coverage and reduced permeability. Together, our study denotes the role of endothelial-ANGPTL4 in regulating cellular metabolism and angiogenic functions of EC.
Collapse
Affiliation(s)
- Balkrishna Chaube
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Kathryn M Citrin
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| | - Mahnaz Sahraei
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Abhishek K Singh
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Diego Saenz de Urturi
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Wen Ding
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Richard W Pierce
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Raaisa Raaisa
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Rebecca Cardone
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Richard Kibbey
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
McLarnon SR, Johnson C, Sun J, Wei Q, Csanyi G, O'Herron P, Marshall B, Giddens P, Sullivan JC, Barrett A, O'Connor PM. Extravasation of Blood and Blood Toxicity Drives Tubular Injury from RBC Trapping in Ischemic AKI. FUNCTION 2023; 4:zqad050. [PMID: 37753180 PMCID: PMC10519276 DOI: 10.1093/function/zqad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Red blood cell (RBC) trapping is common in ischemic acute kidney injury (AKI) and presents as densely packed RBCs that accumulate within and engorge the kidney medullary circulation. In this study, we tested the hypothesis that "RBC trapping directly promotes tubular injury independent of extending ischemia time." Studies were performed on rats. Red blood cell congestion and tubular injury were compared between renal arterial clamping, venous clamping, and venous clamping of blood-free kidneys. Vessels were occluded for either 15 or 45 min with and without reperfusion. We found that RBC trapping in the medullary capillaries occurred rapidly following reperfusion from renal arterial clamping and that this was associated with extravasation of blood from congested vessels, uptake of blood proteins by the tubules, and marked tubular injury. To determine if this injury was due to blood toxicity or an extension of ischemia time, we compared renal venous and arterial clamping without reperfusion. Venous clamping resulted in RBC trapping and marked tubular injury within 45 min of ischemia. Conversely, despite the same ischemia time, RBC trapping and tubular injury were minimal following arterial clamping without reperfusion. Confirming the role of blood toward tubular injury, injury was markedly reduced in blood-free kidneys with venous clamping. Our data demonstrate that RBC trapping results in the rapid extravasation and uptake of blood components by tubular cells, causing toxic tubular injury. Tubular toxicity from extravasation of blood following RBC trapping appears to be a major component of tubular injury in ischemic AKI, which has not previously been recognized.
Collapse
Affiliation(s)
- Sarah R McLarnon
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, 27599, Chapel Hill, NC, USA
| | - Chloe Johnson
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Jingping Sun
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Qingqing Wei
- Department of Anatomy and Cell Biology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Gabor Csanyi
- Department of Pharmacology and Toxicology, Augusta University, 30912, Augusta, GA, USA
| | - Phillip O'Herron
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Brendan Marshall
- Department of Anatomy and Cell Biology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Priya Giddens
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Amanda Barrett
- Department of Pathology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia, Augusta University, 30912, Augusta, GA, USA
| |
Collapse
|
10
|
Garrity R, Arora N, Haque MA, Weis D, Trinh RT, Neerukonda SV, Kumari S, Cortez I, Ubogu EE, Mahalingam R, Tavares-Ferreira D, Price TJ, Kavelaars A, Heijnen CJ, Shepherd AJ. Fibroblast-derived PI16 sustains inflammatory pain via regulation of CD206 + myeloid cells. Brain Behav Immun 2023; 112:220-234. [PMID: 37315702 PMCID: PMC10527931 DOI: 10.1016/j.bbi.2023.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/26/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023] Open
Abstract
Originally identified in fibroblasts, Protease Inhibitor (PI)16 was recently shown to be crucial for the development of neuropathic pain via effects on blood-nerve barrier permeability and leukocyte infiltration, though its impact on inflammatory pain has not been established. Using the complete Freund's Adjuvant inflammatory pain model, we show that Pi16-/- mice are protected against sustained inflammatory pain. Accordingly, intrathecal delivery of a PI16 neutralizing antibody in wild-type mice prevented sustained CFA pain. In contrast to neuropathic pain models, we did not observe any changes in blood-nerve barrier permeability due to PI16 deletion. Instead, Pi16-/- mice display reduced macrophage density in the CFA-injected hindpaw. Furthermore, there was a significant bias toward CD206hi (anti-inflammatory) macrophages in the hindpaw and associated dorsal root ganglia. Following CFA, intrathecal depletion of CD206+ macrophages using mannosylated clodronate liposomes promoted sustained pain in Pi16-/- mice. Similarly, an IL-10 neutralizing antibody also promoted sustained CFA pain in the Pi16-/ when administered intrathecally. Collectively, our results point to fibroblast-derived PI16 mediating substantial differences in macrophage phenotype in the pain neuroaxis under conditions of inflammation. The co-expression of PI16 alongside fibroblast markers in human DRG raise the likelihood that a similar mechanism operates in human inflammatory pain states. Collectively, our findings may have implications for targeting fibroblast-immune cell crosstalk for the treatment of chronic pain.
Collapse
Affiliation(s)
- Rachelle Garrity
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Neha Arora
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Md Areeful Haque
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Drew Weis
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ronnie T Trinh
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sanjay V Neerukonda
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Susmita Kumari
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ibdanelo Cortez
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eroboghene E Ubogu
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, UK
| | - Rajasekaran Mahalingam
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Andrew J Shepherd
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
11
|
Hurtado KA, Janda J, Schnellmann RG. Lasmiditan promotes recovery from acute kidney injury through induction of mitochondrial biogenesis. Am J Physiol Renal Physiol 2023; 324:F56-F63. [PMID: 36326468 PMCID: PMC9762961 DOI: 10.1152/ajprenal.00249.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Acute kidney injury (AKI) involves rapid loss of renal function and occurs in 8-16% of hospitalized patients. AKI can be induced by drugs, sepsis, and ischemia-reperfusion (I/R). Hallmarks of AKI include mitochondrial and microvasculature dysfunction as well as renal tubular injury. There is currently no available therapeutic for AKI. Previously, our group identified that serotonin (5-HT)1F receptor agonism with lasmiditan accelerated endothelial cell recovery and induced mitochondrial biogenesis (MB) in vitro. We hypothesized that lasmiditan, a Federal Drug Administration-approved drug, would induce MB and improve microvascular and renal function in a mouse model of AKI. Male mice were subjected to renal I/R and treated with lasmiditan (0.3 mg/kg) or vehicle beginning 24 h after injury and then daily until euthanasia at 6 or 12 days. Serum creatinine was measured to estimate glomerular filtration rate. The renal cortex was assessed for mitochondrial density, vascular permeability and integrity, tubular damage, and interstitial fibrosis. Lasmiditan increased mitochondrial number (1.4-fold) in renal cortices. At 6 days, serum creatinine decreased 41% in the I/R group and 72% with lasmiditan. At 6 or 12 days, kidney injury molecule-1 increased in the I/R group and decreased 50% with lasmiditan. At 12 days, interstitial fibrosis decreased with lasmiditan by 50% and collagen type 1 by 38%. Evan's blue dye leakage increased 2.5-fold in the I/R group and was restored with lasmiditan. The tight junction proteins zonula occludens-1, claudin-2, and claudin-5 decreased in the I/R group and recovered with lasmiditan. At 6 or 12 days, peroxisome proliferator-activated receptor-γ coactivator-1α and electron transport chain complexes increased only with lasmiditan. In conclusion, lasmiditan treatment beginning AKI induces MB, attenuated vascular and tubular injury, decreased interstitial fibrosis, and lowered serum creatinine. Given that lasmiditan is a Federal Drug Administration-approved drug, these preclinical data support repurposing lasmiditan as a therapeutic for AKI.NEW & NOTEWORTHY AKI pathology involves a rapid decline in kidney function and occurs in 8-16% of hospitalized patients. There is currently no therapeutic for AKI. AKI results in mitochondria dysfunction, microvasculature injury, and loss of renal tubular function. In an I/R-induced AKI mouse model, treatment with the FDA-approved 5-HT1F receptor-selective agonist lasmiditan induced mitochondrial biogenesis, improved vascular integrity, reduced fibrosis, and reduced proximal tubule damage. These data support repurposing lasmiditan for the treatment of AKI.
Collapse
Affiliation(s)
- Kevin A Hurtado
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
- Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
- Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
12
|
Jin H, Luo R, Li J, Zhao H, Ouyang S, Yao Y, Chen D, Ling Z, Zhu W, Chen M, Liao X, Pi J, Huang G. Inhaled platelet vesicle-decoyed biomimetic nanoparticles attenuate inflammatory lung injury. Front Pharmacol 2022; 13:1050224. [PMID: 36523494 PMCID: PMC9745055 DOI: 10.3389/fphar.2022.1050224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/16/2022] [Indexed: 01/04/2024] Open
Abstract
Acute lung injury (ALI) is an inflammatory response which causes serious damages to alveolar epithelia and vasculature, and it still remains high lethality and mortality with no effective treatment. Based on the inflammatory homing of platelets and cell membrane cloaking nanotechnology, in this study we developed a biomimetic anti-inflammation nanoparticle delivery system for ALI treatment. PM@Cur-RV NPs were designed by combining the poly (lactic-co-glycolic acid) nanoparticles (NPs) coated with platelet membrane vesicles (PM) for the purpose of highly targeting delivery of curcumin (Cur) and resveratrol (RV) to inflammatory lungs. PM@Cur-RV NPs showed good biocompatibility and biosafety both in vitro and in vivo. Accumulation of NPs into lung tract was observed after inhaled NPs. Remarkably, the inhalation of PM@Cur-RV NPs effectively inhibited lung vascular injury evidenced by the decreased lung vascular permeability, and the reduced proinflammatory cytokine burden in an ALI mouse model. The analysis of infiltrated macrophages in the lungs showed that the Cur-RV-modulated macrophage polarized towards M2 phenotype and the decreased histone lactylation might contribute to their anti-inflammation effects. Together, this work highlights the potential of inhalation of biomimetic nanoparticle delivery of curcumin and resveratrol for the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Renxing Luo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jianing Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hongxia Zhao
- School of Biomedical and Pharmaceutical Science, Guangdong University of Technology, Guangzhou, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yinlian Yao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Dongyan Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zijie Ling
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Weicong Zhu
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Meijun Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xianping Liao
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
13
|
Fan M, Yang K, Wang X, Zhang X, Xu J, Tu F, Gill PS, Ha T, Williams DL, Li C. LACTATE IMPAIRS VASCULAR PERMEABILITY BY INHIBITING HSPA12B EXPRESSION VIA GPR81-DEPENDENT SIGNALING IN SEPSIS. Shock 2022; 58:304-312. [PMID: 36256626 PMCID: PMC9584042 DOI: 10.1097/shk.0000000000001983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 08/11/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Introduction: Sepsis impaired vascular integrity results in multiple organ failure. Circulating lactate level is positively correlated with sepsis-induced mortality. We investigated whether lactate plays a role in causing endothelial barrier dysfunction in sepsis. Methods: Polymicrobial sepsis was induced in mice by cecal ligation and puncture (CLP). Lactic acid was injected i.p. (pH 6.8, 0.5 g/kg body weight) 6 h after CLP or sham surgery. To elucidate the role of heat shock protein A12B (HSPA12B), wild-type, HSPA12B-transgenic, and endothelial HSPA12B-deficient mice were subjected to CLP or sham surgery. To suppress lactate signaling, 3OBA (120 μM) was injected i.p. 3 h before surgery. Vascular permeability was evaluated with the Evans blue dye penetration assay. Results: We found that administration of lactate elevated CLP-induced vascular permeability. Vascular endothelial cadherin (VE-cadherin), claudin 5, and zonula occluden 1 (ZO-1) play a crucial role in the maintenance of endothelial cell junction and vascular integrity. Lactate administration significantly decreased VE-cadherin, claudin 5, and ZO-1 expression in the heart of septic mice. Our in vitro data showed that lactate (10 mM) treatment disrupted VE-cadherin, claudin 5, and ZO-1 in endothelial cells. Mechanistically, we observed that lactate promoted VE-cadherin endocytosis by reducing the expression of HSPA12B. Overexpression of HSPA12B prevented lactate-induced VE-cadherin disorganization. G protein-coupled receptor 81 (GPR81) is a specific receptor for lactate. Inhibition of GPR81 with its antagonist 3OBA attenuated vascular permeability and reversed HSPA12B expression in septic mice. Conclusions: The present study demonstrated a novel role of lactate in promoting vascular permeability by decreasing VE-cadherin junctions and tight junctions in endothelial cells. The deleterious effects of lactate in vascular hyperpermeability are mediated via HSPA12B- and GPR81-dependent signaling.
Collapse
Affiliation(s)
- Min Fan
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Kun Yang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Xia Zhang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Jingjing Xu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Fei Tu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - P. Spencer Gill
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
14
|
Li Z, He P, Xu Y, Deng Y, Gao Y, Chen SL. In vivo evaluation of a lipopolysaccharide-induced ear vascular leakage model in mice using photoacoustic microscopy. BIOMEDICAL OPTICS EXPRESS 2022; 13:4802-4816. [PMID: 36187238 PMCID: PMC9484413 DOI: 10.1364/boe.471079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 06/16/2023]
Abstract
Sepsis is caused by dysregulated host inflammatory response to infection. During sepsis, early identification and monitoring of vascular leakage are pivotal for improved diagnosis, treatment, and prognosis. However, there is a lack of research on noninvasive observation of inflammation-related vascular leakage. Here, we investigate the use of photoacoustic microscopy (PAM) for in vivo visualization of lipopolysaccharide (LPS)-induced ear vascular leakage in mice using Evans blue (EB) as an indicator. A model combining needle pricking on the mouse ear, topical smearing of LPS on the mouse ear, and intravenous tail injection of EB is developed. Topical application of LPS is expected to induce local vascular leakage in skin. Inflammatory response is first validated by ex vivo histology and enzyme-linked immunosorbent assay. Then, local ear vascular leakage is confirmed by ex vivo measurement of swelling, thickening, and EB leakage. Finally, PAM for in vivo identification and evaluation of early vascular leakage using the model is demonstrated. For PAM, common excitation wavelength of 532 nm is used, and an algorithm is developed to extract quantitative metrics for EB leakage. The results show potential of PAM for noninvasive longitudinal monitoring of peripheral skin vascular leakage, which holds promise for clinical sepsis diagnosis and management.
Collapse
Affiliation(s)
- Zhe Li
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- These authors contributed equally to this work
| | - Pengbo He
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
- These authors contributed equally to this work
| | - Yuqing Xu
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yuxiao Deng
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yuan Gao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sung-Liang Chen
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
- Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, Shanghai 200030, China
- State Key Laboratory of Advanced Optical Communication Systems and Networks, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
15
|
Ivnitsky JJ, Schäfer TV, Rejniuk VL, Vakunenkova OA. Secondary Dysfunction of the Intestinal Barrier in the Pathogenesis of Complications of Acute Poisoning. J EVOL BIOCHEM PHYS+ 2022; 58:1075-1098. [PMID: 36061072 PMCID: PMC9420239 DOI: 10.1134/s0022093022040123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
Abstract
The last decade has been marked by an exponential increase
in the number of publications on the physiological role of the normal
human gut microbiota. The idea of a symbiotic relationship between
the human organism and normal microbiota of its gastrointestinal
tract has been firmly established as an integral part of the current
biomedical paradigm. However, the type of this symbiosis varies
from mutualism to parasitism and depends on the functional state
of the host organism. Damage caused to the organism by external
agents can lead to the emergence of conditionally pathogenic properties
in the normal gut microbiota, mediated by humoral factors and affecting
the outcome of exogenous exposure. Among the substances produced
by symbiotic microbiota, there are an indefinite number of compounds
with systemic toxicity. Some occur in the intestinal chyme in potentially
lethal amounts in the case they enter the bloodstream quickly. The quick
entry of potential toxicants is prevented by the intestinal barrier
(IB), a set of structural elements separating the intestinal chyme
from the blood. Hypothetically, severe damage to the IB caused by
exogenous toxicants can trigger a leakage and subsequent systemic
redistribution of toxic substances of bacterial origin. Until recently,
the impact of such a redistribution on the outcome of acute exogenous
poisoning remained outside the view of toxicology. The present review
addresses causal relationships between the secondary dysfunction
of the IB and complications of acute poisoning. We characterize
acute systemic toxicity of such waste products of the normal gut microflora
as ammonia and endotoxins, and demonstrate their involvement in
the formation of such complications of acute poisoning as shock,
sepsis, cerebral insufficiency and secondary lung injuries. The
principles of assessing the functional state of the IB and the approaches
to its protection in acute poisoning are briefly considered.
Collapse
Affiliation(s)
- Ju. Ju. Ivnitsky
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| | - T. V. Schäfer
- State Scientific Research Test Institute of Military Medicine, Ministry of Defense of the Russian Federation, St. Petersburg, Russia
| | - V. L. Rejniuk
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| | - O. A. Vakunenkova
- Golikov Research Clinical Center of Toxicology, Federal Medical Biological Agency, St. Petersburg, Russia
| |
Collapse
|
16
|
A single-cell survey unveils cellular heterogeneity and sensitive responses in mouse cortices induced by oral exposure to triphenyl phosphate. Arch Toxicol 2022; 96:2545-2557. [PMID: 35752650 DOI: 10.1007/s00204-022-03301-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/07/2022] [Indexed: 11/02/2022]
Abstract
Triphenyl phosphate (TPhP) is a non-halogenated organophosphorus flame retardant, and there is a higher exposure risk in children. TPhP has been found to be neurotoxic upon developmental exposure, yet the specific mechanism remains unclear. To characterize the cellular responses underlying TPhP-induced developmental neurotoxicity, we administered TPhP (0.5, 5 or 50 mg/kg/day) to neonatal mice from postnatal day 10 (P10)-P70. A total of 17,229 cells and 26,338 genes were identified in cortical samples from control and low-dose (the internal doses of metabolite DPhP comparable to human exposure level) groups using single-cell RNA sequencing (scRNA-seq). TPhP exposure led to heterogeneous transcriptional alterations and intercellular crosstalk among neurons, neural stem/progenitor cells (NSPCs), endothelial cells, and immunocytes. Deprivation of NSPCs, loss of mature neurons, and concomitant neuroinflammation mediated by extrinsic and intrinsic immunocytes were found in TPhP-exposed cortices. In addition, we observed blood-brain barrier destruction prior to the anxiety/depression-like neurobehavioral changes. These results reveal the distinctive cellular processes in TPhP's neurodevelopmental toxicity and uncover that the impeded neurogenesis, disrupted vascular barrier, and concomitant neuroinflammation are the sensitive responses to TPhP exposure. Our study paves the way for the application of scRNA-seq in toxicity assessments for emerging neurotoxic pollutants.
Collapse
|
17
|
Khamissi FZ, Ning L, Kefaloyianni E, Dun H, Arthanarisami A, Keller A, Atkinson JJ, Li W, Wong B, Dietmann S, Lavine K, Kreisel D, Herrlich A. Identification of kidney injury released circulating osteopontin as causal agent of respiratory failure. SCIENCE ADVANCES 2022; 8:eabm5900. [PMID: 35213222 PMCID: PMC8880785 DOI: 10.1126/sciadv.abm5900] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/30/2021] [Indexed: 05/08/2023]
Abstract
Tissue injury can drive secondary organ injury; however, mechanisms and mediators are not well understood. To identify interorgan cross-talk mediators, we used acute kidney injury (AKI)-induced acute lung injury (ALI) as a clinically important example. Using kidney and lung single-cell RNA sequencing after AKI in mice followed by ligand-receptor pairing analysis across organs, kidney ligands to lung receptors, we identify kidney-released circulating osteopontin (OPN) as a novel AKI-ALI mediator. OPN release from kidney tubule cells triggered lung endothelial leakage, inflammation, and respiratory failure. Pharmacological or genetic OPN inhibition prevented AKI-ALI. Transplantation of ischemic wt kidneys caused AKI-ALI, but not of ischemic OPN-global knockout kidneys, identifying kidney-released OPN as necessary interorgan signal to cause AKI-ALI. We show that OPN serum levels are elevated in patients with AKI and correlate with kidney injury. Our results demonstrate feasibility of using ligand-receptor analysis across organs to identify interorgan cross-talk mediators and may have important therapeutic implications in human AKI-ALI and multiorgan failure.
Collapse
Affiliation(s)
| | | | | | - Hao Dun
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | | | - Amy Keller
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey J. Atkinson
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Wenjun Li
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Brian Wong
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Sabine Dietmann
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Kory Lavine
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Daniel Kreisel
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | | |
Collapse
|
18
|
Tumor Necrosis Factor-α Mediates Lung Injury in the Early Phase of Endotoxemia. Pharmaceuticals (Basel) 2022; 15:ph15030287. [PMID: 35337084 PMCID: PMC8953981 DOI: 10.3390/ph15030287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 11/23/2022] Open
Abstract
Endotoxemia induces lung injury. We assessed the therapeutic efficacy between triple cytokine (tumor necrosis factor-α [TNF-α], interleukin-1β [IL-1β], and IL-6) inhibition (mediated by KCF18 peptide) and single cytokine (TNF-α) inhibition (mediated by SEM18 peptide) on alleviating lung injury in the early phase of endotoxemia. Mice receiving endotoxin (Endo group), endotoxin plus KCF18 (EKCF group), or endotoxin plus SEM18 (ESEM) were monitored and euthanized at 24 h after endotoxin. Our data demonstrated altered lung function (decreases in tidal volume, minute ventilation, and dynamic compliance; and by contrast, increases in airway resistance and end expiration work) and histology (increases in injury scores, leukocyte infiltration, vascular permeability, and tissue water content) in the Endo group with significant protection observed in the EKCF and ESEM groups (all p < 0.05). Levels of inflammation (macrophage activation and cytokine upregulations), oxidation (lipid peroxidation), necroptosis, pyroptosis, and apoptosis in EKCF and ESEM groups were comparable and all were significantly lower than in the Endo group (all p < 0.05). These data demonstrate that single cytokine TNF-α inhibition can achieve therapeutic effects similar to triple cytokines TNF-α, IL-1β, and IL-6 inhibition on alleviating endotoxin-induced lung injury, indicating that TNF-α is the major cytokine in mediating lung injury in the early phase of endotoxemia.
Collapse
|
19
|
Chuang DJ, Pethaperumal S, Siwakoti B, Chien HJ, Cheng CF, Hung SC, Lien TS, Sun DS, Chang HH. Activating Transcription Factor 3 Protects against Restraint Stress-Induced Gastrointestinal Injury in Mice. Cells 2021; 10:3530. [PMID: 34944038 PMCID: PMC8700235 DOI: 10.3390/cells10123530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/07/2021] [Accepted: 12/11/2021] [Indexed: 12/11/2022] Open
Abstract
Psychological stress increases the risk of gastrointestinal (GI) tract diseases, which involve bidirectional communication of the GI and nerves systems. Acute stress leads to GI ulcers; however, the mechanism of the native cellular protection pathway, which safeguards tissue integrality and maintains GI homeostasis, remains to be investigated. In a mouse model of this study, restraint stress induced GI leakage, abnormal tight junction protein expression, and cell death of gut epithelial cells. The expression of activating transcription factor 3 (ATF3), a stress-responsive transcription factor, is upregulated in the GI tissues of stressed animals. ATF3-deficient mice displayed an exacerbated phenotype of GI injuries. These results suggested that, in response to stress, ATF3 is part of the native cellular protective pathway in the GI system, which could be a molecular target for managing psychological stress-induced GI tract diseases.
Collapse
Affiliation(s)
- Dun-Jie Chuang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-J.C.); (S.P.); (B.S.); (T.-S.L.); (D.-S.S.)
| | - Subhashree Pethaperumal
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-J.C.); (S.P.); (B.S.); (T.-S.L.); (D.-S.S.)
| | - Bijaya Siwakoti
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-J.C.); (S.P.); (B.S.); (T.-S.L.); (D.-S.S.)
| | - Hung-Jen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 300, Taiwan;
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan;
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Shih-Che Hung
- Institute of Medical Sciences, Tzu-Chi University, Hualien 970, Taiwan;
| | - Te-Sheng Lien
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-J.C.); (S.P.); (B.S.); (T.-S.L.); (D.-S.S.)
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-J.C.); (S.P.); (B.S.); (T.-S.L.); (D.-S.S.)
- Institute of Medical Sciences, Tzu-Chi University, Hualien 970, Taiwan;
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (D.-J.C.); (S.P.); (B.S.); (T.-S.L.); (D.-S.S.)
- Institute of Medical Sciences, Tzu-Chi University, Hualien 970, Taiwan;
| |
Collapse
|
20
|
Liu L, Shao Y, Zhang Y, Yang Y, Huang J, Li L, Sun R, Zhou Y, Su Y, Sun B. Neutrophil-derived heparin binding protein triggers vascular leakage and synergizes with myeloperoxidase at the early stage of severe burns (With video). BURNS & TRAUMA 2021; 9:tkab030. [PMID: 34646891 PMCID: PMC8499692 DOI: 10.1093/burnst/tkab030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/13/2021] [Indexed: 11/13/2022]
Abstract
Background Burn shock caused by vascular leakage is one of the main causes of high mortality in severe burn injury. However, the pathophysiological mechanism of vascular leakage is still unclear. The purpose of this study was to explore the molecular mechanism of vascular leakage in the early stage of severe burn and provide a new target for the treatment of severe burns. Methods Neutrophils were isolated from human peripheral blood by magnetic beads sorting. ELISA was used to detect neutrophil-derived granule proteins and glycocalyx injury products in plasma. The vascular leakage and neutrophil movement were assessed by in vivo laser confocal imaging in mice, and high-quality video were provided.. Adhesion-related molecules were investigated by qRT-PCR. The damage to glycocalyx of mice vascular endothelial cells was observed by transmission electron microscope and scanning electron microscope. Proteomic analysis, flow cytometry and immunofluorescence were used to further study the relationship between human peripheral blood neutrophil-derived hypochlorite (HOCl) and CD44 of human vascular endothelial cells. Results In this study, we found that rapidly increasing activated neutrophils secrete heparin binding protein (HBP) and myeloperoxidase (MPO) after severe burn injury. Increased HBP triggers vascular leakage with synergy of MPO, results in systemic edema and burn shock. Furthermore, we found that the MPO catalytic product HOCl but not MPO triggers CD44 extracellular domain shedding from vascular endothelial cells to damage the glycocalyx. Damage to the glycocalyx results in firm adhesion of neutrophils and increases vascular leakage. However, MPO inhibitors partially protect the glycocalyx of vascular endothelial cells. The combination of HBP and MPO inhibitors markedly reduces vascular leakage and systemic edema in the early stage of severe burns. Conclusions Taken together, these data reveal that neutrophil-derived HBP and MPO play an important synergies role in triggering vascular leakage at the early stage of severe burns. Targeted intervention in these two biomolecules may introduce new strategies for helping to reduce large amount of fluid loss and subsequent burn shock.
Collapse
Affiliation(s)
- Lu Liu
- School of Medicine, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Yiming Shao
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Yixuan Zhang
- School of Medicine, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Yunxi Yang
- School of Medicine, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Jiamin Huang
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Linbin Li
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Ran Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Yuying Zhou
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Yicheng Su
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| | - Bingwei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, Jiangsu Province, China
| |
Collapse
|
21
|
Berber E, Sumbria D, Newkirk KM, Rouse BT. Inhibiting Glucose Metabolism Results in Herpes Simplex Encephalitis. THE JOURNAL OF IMMUNOLOGY 2021; 207:1824-1835. [PMID: 34470854 DOI: 10.4049/jimmunol.2100453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/31/2021] [Indexed: 11/19/2022]
Abstract
This report evaluates how HSV enters the brain to cause herpes simplex encephalitis following infection at a peripheral site. We demonstrate that encephalitis regularly occurred when BALB/c mice were infected with HSV and treated daily with 2-deoxy-d-glucose (2DG), which inhibits glucose use via the glycolysis pathway. The outcome of infection in the trigeminal ganglion (TG), the site to which the virus spreads, replicates, and establishes latency, showed marked differences in viral and cellular events between treated and untreated animals. In control-untreated mice, the replicating virus was present only during early time points, whereas in 2DG recipients, replicating virus remained for the 9-d observation period. This outcome correlated with significantly reduced numbers of innate inflammatory cells as well as T cells in 2DG-treated animals. Moreover, T cells in the TG of treated animals were less activated and contained a smaller fraction of expressed IFN-γ production compared with untreated controls. The breakdown of latency was accelerated when cultures of TG cells taken from mice with established HSV latency were cultured in the presence of 2DG. Taken together, the results of both in vivo and in vitro investigations demonstrate that the overall effects of 2DG therapy impaired the protective effects of one or more inflammatory cell types in the TG that normally function to control productive infection and prevent spread of virus to the brain.
Collapse
Affiliation(s)
- Engin Berber
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN; and.,Department of Virology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Deepak Sumbria
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN; and
| | - Kim M Newkirk
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN; and
| | - Barry T Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN; and
| |
Collapse
|
22
|
Alves-Lopes R, Montezano AC, Neves KB, Harvey A, Rios FJ, Skiba DS, Arendse LB, Guzik TJ, Graham D, Poglitsch M, Sturrock E, Touyz RM. Selective Inhibition of the C-Domain of ACE (Angiotensin-Converting Enzyme) Combined With Inhibition of NEP (Neprilysin): A Potential New Therapy for Hypertension. Hypertension 2021; 78:604-616. [PMID: 34304582 PMCID: PMC8357049 DOI: 10.1161/hypertensionaha.121.17041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Augusto C. Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Karla B. Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Adam Harvey
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Francisco J. Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Dominik S. Skiba
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Lauren B. Arendse
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Biochemistry, University of Cape Town, South Africa (L.B.A., E.S.)
| | - Tomasz J. Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| | | | - Edward Sturrock
- Institute of Infectious Disease and Molecular Medicine and Division of Medical Biochemistry, University of Cape Town, South Africa (L.B.A., E.S.)
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (R.A.-L., A.C.M., K.B.N., A.H., F.J.R., D.S.S., T.J.G., D.G., R.M.T.)
| |
Collapse
|
23
|
Ye H, Zhang Y, Huang Y, Li B, Cao R, Dai L, Huang B, Tian P, Li L, Han Y. Bivalirudin Attenuates Thrombin-Induced Endothelial Hyperpermeability via S1P/S1PR2 Category: Original Articles. Front Pharmacol 2021; 12:721200. [PMID: 34413778 PMCID: PMC8369898 DOI: 10.3389/fphar.2021.721200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/23/2021] [Indexed: 12/02/2022] Open
Abstract
Aims: To explore the role of the Sphingosine 1-Phosphate (S1P)/Receptor2 (S1PR2) pathway in thrombin-induced hyperpermeability (TIP) and to test whether bivalirudin can reverse TIP via the S1P-S1PRs pathway. Methods and Results: Using western blot, we demonstrated that Human umbilical vein endothelial cells (HUVECs) that were cultured with 2 U/ml thrombin showed significantly increased S1PR2 expression while S1PR1and three kept unchanged. Such increment was attenuated by JTE-013 pretreatment and by presence of bivalirudin. Exposure of 2 U/ml of thrombin brought a higher level of S1P both intracellularly and extracellularly within the HUVECs by using ELISA detecting. Thrombin induced S1P and S1PR2 increment was restored by usage of PF543 and bivalirudin. Bivalirudin alone did not influenced the level of S1P and S1PR1,2, and S1PR3 compare to control group. As a surrogate of cytoskeleton morphology, phalloidin staining and immunofluorescence imaging were used. Blurry cell edges and intercellular vacuoles or spaces were observed along thrombin-exposed HUVECs. Presence of JTE-013 and bivalirudin attenuated such thrombin-induced permeability morphological change and presence of heparin failed to show the protective effect. Transwell chamber assay and probe assay were used to measure and compare endothelial permeability in vitro. An increased TIP was observed in HUVECs cultured with thrombin, and coculture with bivalirudin, but not heparin, alleviated this increase. JTE-013 treatment yielded to similar TIP alleviating effect. In vivo, an Evans blue assay was used to test subcutaneous and organ microvascular permeability after the treatment of saline only, thrombin + saline, thrombin + bivalirudin, thrombin + heparin or thrombin + JTE-013. Increased subcutaneous and organ tissue permeability after thrombin treatment was observed in thrombin + saline and thrombin + heparin groups while treatment of bivalirudin and JTE-013 absent this effect. Conclusion: S1P/S1PR2 mediates TIP by impairing vascular endothelial barrier function. Unlike heparin, bivalirudin effectively blocked TIP by inhibiting the thrombin-induced S1P increment and S1PR2 expression, suggesting the novel endothelial protective effect of bivalirudin under pathological procoagulant circumstance.
Collapse
Affiliation(s)
- Haowen Ye
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yizhi Zhang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Huang
- Department of Pediatrics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Biao Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Ruhao Cao
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Libing Dai
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Bin Huang
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Pingge Tian
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
24
|
Lin Y, Xiao S, Yao W, Lv Z, Tang Y, Zhang Y, Chen L. Molecular photoacoustic imaging for early diagnosis and treatment monitoring of rheumatoid arthritis in a mouse model. Am J Transl Res 2021; 13:8873-8884. [PMID: 34540001 PMCID: PMC8430181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/11/2021] [Indexed: 06/13/2023]
Abstract
Rheumatoid arthritis (RA) is a progressive inflammatory joint disease. Early diagnosis is critical for timely therapeutic intervention. However, it lacks effective diagnostic methods capable of detecting disease progression in its early stage and evaluating treatment efficacy in clinics. Photoacoustic (PA) molecular imaging is a novel imaging modality that can detect in the early stage of disease and continuously monitor its progression. In this study, Evans blue (EB) was used as a PA contrast agent to detect the angiogenesis and microcirculation dysfunction in RA joint. In collagen-induced arthritis (CIA) mouse model, a distinct increase of PA signal was detected early at 2 weeks, with significant higher PA signal intensities from the RA joints compared to the normal joints. More importantly, we detected an increasing trend of PA signal intensity week by week post CIA induction, demonstrating the potential of EB-enhanced PA imaging in monitoring the development of RA. However, joint damage was silent in the X-ray at 2 weeks post CIA induction, which suggested the superiority of PA imaging in RA early detection. In addition, striking decrease of PA signal intensities in the RA joints was observed after administration with etanercept compared with the untreated RA joints. The signal changes exhibited by PA imaging were confirmed by clinical observation and histological examinations. This study demonstrated the promising use of EB-enhanced PA imaging for the early diagnosis and its feasibility for RA treatment monitoring.
Collapse
Affiliation(s)
- Yimu Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Shuyi Xiao
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang UniversityHangzhou 310016, P. R. China
| | - Wei Yao
- Department of Respiratory Medicine, Huzhou First People’s HospitalHuzhou 313000, P. R. China
| | - Zhuang Lv
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications (NUPT)Nanjing 210023, P. R. China
| | - Yufu Tang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech)Nanjing 211816, P. R. China
| | - Yu Zhang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Liang Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| |
Collapse
|
25
|
Retamal JS, Grace MS, Dill LK, Ramirez-Garcia P, Peng S, Gondin AB, Bennetts F, Alvi S, Rajasekhar P, Almazi JG, Carbone SE, Bunnett NW, Davis TP, Veldhuis NA, Poole DP, McIntyre P. Serotonin-induced vascular permeability is mediated by transient receptor potential vanilloid 4 in the airways and upper gastrointestinal tract of mice. J Transl Med 2021; 101:851-864. [PMID: 33859334 PMCID: PMC8047529 DOI: 10.1038/s41374-021-00593-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 01/07/2023] Open
Abstract
Endothelial and epithelial cells form physical barriers that modulate the exchange of fluid and molecules. The integrity of these barriers can be influenced by signaling through G protein-coupled receptors (GPCRs) and ion channels. Serotonin (5-HT) is an important vasoactive mediator of tissue edema and inflammation. However, the mechanisms that drive 5-HT-induced plasma extravasation are poorly defined. The Transient Receptor Potential Vanilloid 4 (TRPV4) ion channel is an established enhancer of signaling by GPCRs that promote inflammation and endothelial barrier disruption. Here, we investigated the role of TRPV4 in 5-HT-induced plasma extravasation using pharmacological and genetic approaches. Activation of either TRPV4 or 5-HT receptors promoted significant plasma extravasation in the airway and upper gastrointestinal tract of mice. 5-HT-mediated extravasation was significantly reduced by pharmacological inhibition of the 5-HT2A receptor subtype, or with antagonism or deletion of TRPV4, consistent with functional interaction between 5-HT receptors and TRPV4. Inhibition of receptors for the neuropeptides substance P (SP) or calcitonin gene-related peptide (CGRP) diminished 5-HT-induced plasma extravasation. Supporting studies assessing treatment of HUVEC with 5-HT, CGRP, or SP was associated with ERK phosphorylation. Exposure to the TRPV4 activator GSK1016790A, but not 5-HT, increased intracellular Ca2+ in these cells. However, 5-HT pre-treatment enhanced GSK1016790A-mediated Ca2+ signaling, consistent with sensitization of TRPV4. The functional interaction was further characterized in HEK293 cells expressing 5-HT2A to reveal that TRPV4 enhances the duration of 5-HT-evoked Ca2+ signaling through a PLA2 and PKC-dependent mechanism. In summary, this study demonstrates that TRPV4 contributes to 5-HT2A-induced plasma extravasation in the airways and upper GI tract, with evidence supporting a mechanism of action involving SP and CGRP release.
Collapse
Affiliation(s)
- Jeffri S Retamal
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Megan S Grace
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
- Department of Physiology, School of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Clinical Medicine, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Larissa K Dill
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Paulina Ramirez-Garcia
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Scott Peng
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Felix Bennetts
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Sadia Alvi
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Pradeep Rajasekhar
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Juhura G Almazi
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
| | - Simona E Carbone
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Nigel W Bunnett
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia.
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia.
| | - Peter McIntyre
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| |
Collapse
|
26
|
Proença-Assunção JDC, Farias-de-França AP, Tribuiani N, Cogo JC, Collaço RDC, Randazzo-Moura P, Consonni SR, Chaud MV, Dos Santos CA, Oshima-Franco Y. The Influence of Silver Nanoparticles Against Toxic Effects of Philodryas olfersii Venom. Int J Nanomedicine 2021; 16:3555-3564. [PMID: 34079248 PMCID: PMC8164871 DOI: 10.2147/ijn.s293366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/27/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose A silver nanoparticle obtained by reducing salts with solid dispersion of curcumin (130 nm, 0.081 mg mL−1) was used to counteract against the toxic – edematogenic, myotoxic, and neurotoxic – effects of Philodryas olfersii venom. Methods The edematogenic effect was evaluated by plasma extravasation in rat dorsal skin after injection of 50 µg per site of venom alone or preincubated with 1, 10, and 100 µL of AgNPs; the myotoxicity was evaluated by measuring the creatine kinase released into the organ-bath before the treatment and at the end of each experiment; and neurotoxicity was evaluated in chick biventer cervicis using the conventional myographic technique, face to the exogenous acetylcholine (ACh) and potassium chloride (KCl) added into the bath before the treatment and after each experiment. Preliminarily, a concentration-response curve of AgNPs was carried out to select the concentration to be used for neutralizing assays, which consists of neutralizing the venom-induced neuromuscular paralysis and edema by preincubating AgNPs with venom for 30 min. Results The P. olfersii venom-induced edema (n=6) and a complete neuromuscular blockade (n=4) that includes the total and unrecovered block of ACh and KCl contractures. AgNPs produced a concentration-dependent decrease the venom-induced edema (n=6) from 223.3% to 134.4% and to 100.5% after 10 and 100 µL AgNPs-preincubation, respectively. The preincubation of venom with AgNPs (1 µL; n=6) was able to maintain 46.5 ± 10.9% of neuromuscular response under indirect stimuli, 39.2 ± 9.7% of extrinsic nicotinic receptors functioning in absence of electrical stimulus and 28.3 ± 8.1% of responsiveness to potassium on the sarcolemmal membrane. The CK release was not affected by any experimental protocol which was like control. Conclusion AgNPs interact with constituents of P. olfersii venom responsible for the edema-forming activity and neuromuscular blockade, but not on the sarcolemma membrane-acting constituents. The protective effect of the studied AgNPs on avian preparation points out to molecular targets as intrinsic and extrinsic nicotinic receptors.
Collapse
Affiliation(s)
| | | | - Natalia Tribuiani
- Post-Graduate Program in Pharmaceutical Sciences, University of Sorocaba (Uniso), Sorocaba, SP, Brazil
| | - Jose Carlos Cogo
- Bioengineering and Biomedical Engineering Programs, Technological and Scientific Institute, Brazil University, São Paulo, Brazil
| | - Rita de Cássia Collaço
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Priscila Randazzo-Moura
- Department of Surgery, Pontifícia Universidade Católica De São Paulo (PUCSP), Sorocaba, SP, Brazil
| | - Sílvio Roberto Consonni
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Marco Vinicius Chaud
- Post-Graduate Program in Pharmaceutical Sciences, University of Sorocaba (Uniso), Sorocaba, SP, Brazil
| | | | - Yoko Oshima-Franco
- Post-Graduate Program in Pharmaceutical Sciences, University of Sorocaba (Uniso), Sorocaba, SP, Brazil
| |
Collapse
|
27
|
Shih HJ, Chang CY, Chiang M, Le VL, Hsu HJ, Huang CJ. Simultaneous Inhibition of Three Major Cytokines and Its Therapeutic Effects: A Peptide-Based Novel Therapy against Endotoxemia in Mice. J Pers Med 2021; 11:jpm11050436. [PMID: 34065201 PMCID: PMC8161041 DOI: 10.3390/jpm11050436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Three major cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6, mediate endotoxemia-induced liver injury. With the similar structures to the binding domains of the three cytokines to their cognate receptors, the novel peptide KCF18 can simultaneously inhibit TNF-α, IL-1β, and IL-6. We elucidated whether KCF18 can alleviate injury of liver in endotoxemic mice. Adult male mice (BALB/cJ) were intraperitoneally (i.p.) administered lipopolysaccharide (LPS, 15 mg/kg; LPS group) or LPS with KCF18 (LKCF group). Mice in the LKCF group received KCF18 (i.p.) at 2 h (0.6 mg/kg), 4 h (0.3 mg/kg), 6 h (0.3 mg/kg), and 8 h (0.3mg/kg) after LPS administration. Mice were sacrificed after receiving LPS for 24 h. Our results indicated that the binding levels of the three cytokines to their cognate receptors in liver tissues in the LKCF group were significantly lower than those in the LPS group (all p < 0.05). The liver injury level, as measured by performing functional and histological analyses and by determining the tissue water content and vascular permeability (all p < 0.05), was significantly lower in the LKCF group than in the LPS group. Similarly, the levels of inflammation (macrophage activation, cytokine upregulation, and leukocyte infiltration), oxidation, necroptosis, pyroptosis, and apoptosis (all p < 0.05) in liver tissues in the LKCF group were significantly lower than those in the LPS group. In conclusion, the KCF18 peptide–based simultaneous inhibition of TNF-α, IL-1β, and IL-6 can alleviate liver injury in mice with endotoxemia.
Collapse
Affiliation(s)
- Hung-Jen Shih
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Department of Urology, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Chao-Yuan Chang
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Milton Chiang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (M.C.); (V.L.L.)
| | - Van Long Le
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (M.C.); (V.L.L.)
- Department of Anesthesiology and Critical Care, Hue University of Medicine and Pharmacy, Hue City 52000, Vietnam
| | - Hao-Jen Hsu
- Department of Life Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Correspondence: (H.-J.H.); (C.-J.H.)
| | - Chun-Jen Huang
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (M.C.); (V.L.L.)
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (H.-J.H.); (C.-J.H.)
| |
Collapse
|
28
|
Ghaghada KB, Ren P, Devkota L, Starosolski Z, Zhang C, Vela D, Stupin IV, Tanifum EA, Annapragada AV, Shen YH, LeMaire SA. Early Detection of Aortic Degeneration in a Mouse Model of Sporadic Aortic Aneurysm and Dissection Using Nanoparticle Contrast-Enhanced Computed Tomography. Arterioscler Thromb Vasc Biol 2021; 41:1534-1548. [PMID: 33535789 PMCID: PMC7990703 DOI: 10.1161/atvbaha.120.315210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ketan B Ghaghada
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston (K.B.G., L.D., Z.S., I.V.S., E.A.T., A.V.A.)
- Department of Radiology (K.B.G., Z.S., E.A.T., A.V.A.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (K.B.G., A.V.A., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
| | - Pingping Ren
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (P.R., C.Z., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
| | - Laxman Devkota
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston (K.B.G., L.D., Z.S., I.V.S., E.A.T., A.V.A.)
- Department of Pediatrics, Section of Hematology-Oncology (L.D.), Baylor College of Medicine, Houston, TX
| | - Zbigniew Starosolski
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston (K.B.G., L.D., Z.S., I.V.S., E.A.T., A.V.A.)
- Department of Radiology (K.B.G., Z.S., E.A.T., A.V.A.), Baylor College of Medicine, Houston, TX
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (P.R., C.Z., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
| | - Deborah Vela
- Department of Cardiovascular Pathology Research (D.V.), Texas Heart Institute, Houston
| | - Igor V Stupin
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston (K.B.G., L.D., Z.S., I.V.S., E.A.T., A.V.A.)
| | - Eric A Tanifum
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston (K.B.G., L.D., Z.S., I.V.S., E.A.T., A.V.A.)
- Department of Radiology (K.B.G., Z.S., E.A.T., A.V.A.), Baylor College of Medicine, Houston, TX
| | - Ananth V Annapragada
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston (K.B.G., L.D., Z.S., I.V.S., E.A.T., A.V.A.)
- Department of Radiology (K.B.G., Z.S., E.A.T., A.V.A.), Baylor College of Medicine, Houston, TX
- Cardiovascular Research Institute (K.B.G., A.V.A., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
| | - Ying H Shen
- Cardiovascular Research Institute (K.B.G., A.V.A., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (P.R., C.Z., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (Y.H.S., S.A.L.), Texas Heart Institute, Houston
| | - Scott A LeMaire
- Cardiovascular Research Institute (K.B.G., A.V.A., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (P.R., C.Z., Y.H.S., S.A.L.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (Y.H.S., S.A.L.), Texas Heart Institute, Houston
| |
Collapse
|
29
|
Honeycutt SE, O'Brien LL. Injection of Evans blue dye to fluorescently label and image intact vasculature. Biotechniques 2021; 70:181-185. [PMID: 33337254 PMCID: PMC7983036 DOI: 10.2144/btn-2020-0152] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022] Open
Abstract
Blood vessels perform critical functions in both health and disease. Understanding how vessels form, pattern and respond to damage is essential. However, labeling and imaging the vasculature to ascertain these properties can be difficult and time-consuming. Here, the authors present a novel methodology for rapidly and efficiently labeling whole vascular networks in vivo by exploiting the fluorescent properties of Evans blue. By combining the labeling with fluorescence microscopy, this method enables visualization of whole tissue vasculature for a fraction of the time and cost compared with traditional methods.
Collapse
Affiliation(s)
- Samuel E Honeycutt
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lori L O'Brien
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
30
|
Boerner K, Luissint AC, Parkos CA. Functional Assessment of Intestinal Permeability and Neutrophil Transepithelial Migration in Mice using a Standardized Intestinal Loop Model. J Vis Exp 2021. [PMID: 33645571 PMCID: PMC11404721 DOI: 10.3791/62093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The intestinal mucosa is lined by a single layer of epithelial cells that forms a dynamic barrier allowing paracellular transport of nutrients and water while preventing passage of luminal bacteria and exogenous substances. A breach of this layer results in increased permeability to luminal contents and recruitment of immune cells, both of which are hallmarks of pathologic states in the gut including inflammatory bowel disease (IBD). Mechanisms regulating epithelial barrier function and transepithelial migration (TEpM) of polymorphonuclear neutrophils (PMN) are incompletely understood due to the lack of experimental in vivo methods allowing quantitative analyses. Here, we describe a robust murine experimental model that employs an exteriorized intestinal segment of either ileum or proximal colon. The exteriorized intestinal loop (iLoop) is fully vascularized and offers physiological advantages over ex vivo chamber-based approaches commonly used to study permeability and PMN migration across epithelial cell monolayers. We demonstrate two applications of this model in detail: (1) quantitative measurement of intestinal permeability through detection of fluorescence-labeled dextrans in serum after intraluminal injection, (2) quantitative assessment of migrated PMN across the intestinal epithelium into the gut lumen after intraluminal introduction of chemoattractants. We demonstrate feasibility of this model and provide results utilizing the iLoop in mice lacking the epithelial tight junction-associated protein JAM-A compared to controls. JAM-A has been shown to regulate epithelial barrier function as well as PMN TEpM during inflammatory responses. Our results using the iLoop confirm previous studies and highlight the importance of JAM-A in regulation of intestinal permeability and PMN TEpM in vivo during homeostasis and disease. The iLoop model provides a highly standardized method for reproducible in vivo studies of intestinal homeostasis and inflammation and will significantly enhance understanding of intestinal barrier function and mucosal inflammation in diseases such as IBD.
Collapse
Affiliation(s)
- Kevin Boerner
- Department of Pathology, University of Michigan, Ann Arbor
| | | | | |
Collapse
|
31
|
Jackson MA, Patel SS, Yu F, Cottam MA, Glass EB, Hoogenboezem EN, Fletcher RB, Dollinger BR, Patil P, Liu DD, Kelly IB, Bedingfield SK, King AR, Miles RE, Hasty AM, Giorgio TD, Duvall CL. Kupffer cell release of platelet activating factor drives dose limiting toxicities of nucleic acid nanocarriers. Biomaterials 2021; 268:120528. [PMID: 33285438 PMCID: PMC7856291 DOI: 10.1016/j.biomaterials.2020.120528] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/08/2020] [Accepted: 11/04/2020] [Indexed: 01/06/2023]
Abstract
This work establishes that Kupffer cell release of platelet activating factor (PAF), a lipidic molecule with pro-inflammatory and vasoactive signaling properties, dictates dose-limiting siRNA nanocarrier-associated toxicities. High-dose intravenous injection of siRNA-polymer nano-polyplexes (si-NPs) elicited acute, shock-like symptoms in mice, associated with increased plasma PAF and consequently reduced PAF acetylhydrolase (PAF-AH) activity. These symptoms were completely prevented by prophylactic PAF receptor inhibition or Kupffer cell depletion. Assessment of varied si-NP chemistries confirmed that toxicity level correlated to relative uptake of the carrier by liver Kupffer cells and that this toxicity mechanism is dependent on carrier endosome disruptive function. 4T1 tumor-bearing mice, which exhibit increased circulating leukocytes, displayed greater sensitivity to these toxicities. PAF-mediated toxicities were generalizable to commercial delivery reagent in vivo-jetPEI® and an MC3 lipid formulation matched to an FDA-approved nanomedicine. These collective results establish Kupffer cell release of PAF as a key mediator of siRNA nanocarrier toxicity and identify PAFR inhibition as an effective strategy to increase siRNA nanocarrier tolerated dose.
Collapse
Affiliation(s)
- Meredith A Jackson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Evan B Glass
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Ella N Hoogenboezem
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Bryan R Dollinger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Danielle D Liu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Isom B Kelly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Sean K Bedingfield
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Allyson R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Rachel E Miles
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Alyssa M Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, 37212, USA
| | - Todd D Giorgio
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
32
|
Reventun P, Sanchez-Esteban S, Cook A, Cuadrado I, Roza C, Moreno-Gomez-Toledano R, Muñoz C, Zaragoza C, Bosch RJ, Saura M. Bisphenol A induces coronary endothelial cell necroptosis by activating RIP3/CamKII dependent pathway. Sci Rep 2020; 10:4190. [PMID: 32144343 PMCID: PMC7060177 DOI: 10.1038/s41598-020-61014-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023] Open
Abstract
Epidemiological studies link long term exposure to xenoestrogen Bisphenol-A to adverse cardiovascular effects. Our previous results show that BPA induces hypertension by a mechanism involving CamKII activation and increased redox stress caused by eNOS uncoupling. Recently, CamKII sustained activation has been recognized as a central mediator of programmed cell death in cardiovascular diseases, including necroptosis. However, the role of necroptosis in cardiac response to BPA had not yet been explored. Mice exposed to BPA for 16 weeks showed altered heart function, electrical conduction, and increased blood pressure. Besides, a stress test showed ST-segment depression, indicative of cardiac ischemia. The hearts exhibited cardiac hypertrophy and reduced vascularization, interstitial edema, and large hemorrhagic foci accompanied by fibrinogen deposits. BPA initiated a cardiac inflammatory response, up-regulation of M1 macrophage polarization, and increased oxidative stress, coinciding with the increased expression of CamKII and the necroptotic effector RIP3. In addition, cell death was especially evident in coronary endothelial cells within hemorrhagic areas, and Evans blue extravasation indicated a vascular leak in response to Bisphenol-A. Consistent with the in vivo findings, BPA increased the necroptosis/apoptosis ratio, the expression of RIP3, and CamKII activation in endothelial cells. Necrostatin-1, an inhibitor of necroptosis, alleviated BPA induced cardiac dysfunction and prevented the inflammatory and hemorrhagic response in mice. Mechanistically, silencing of RIP3 reversed BPA-induced necroptosis and CamKII activation in endothelial cells, while inhibition of CamKII activation by KN-93 had no effect on RIP3 expression but decreased necroptotic cell death suggesting that BPA induced necroptosis is mediated by a RIP 3/CamKII dependent pathway. Our results reveal a novel pathogenic role of BPA on the coronary circulation. BPA induces endothelial cell necroptosis, promotes the weakening of coronary vascular wall, which caused internal ventricular hemorrhages, delaying the reparative process and ultimately leading to cardiac dysfunction.
Collapse
Affiliation(s)
- P Reventun
- Biology systems Dpt, University Alcalá (UAH), Madrid, Spain
| | | | - A Cook
- Biology systems Dpt, University Alcalá (UAH), Madrid, Spain
| | - I Cuadrado
- Pharmacology, Pharmacognosy and Botanics Dpt, Complutense University (UCM), Madrid, Spain
| | - C Roza
- Biology systems Dpt, University Alcalá (UAH), Madrid, Spain
| | | | - C Muñoz
- Biology systems Dpt, University Alcalá (UAH), Madrid, Spain
| | - C Zaragoza
- Joint Unit of Cardiovascular Research University Francisco de Vitoria and Hospital Ramon y Cajal, Madrid, Spain
| | - R J Bosch
- Biology systems Dpt, University Alcalá (UAH), Madrid, Spain
| | - M Saura
- Biology systems Dpt, University Alcalá (UAH), Madrid, Spain.
| |
Collapse
|